1
|
Jurcau MC, Jurcau A, Cristian A, Hogea VO, Diaconu RG, Nunkoo VS. Inflammaging and Brain Aging. Int J Mol Sci 2024; 25:10535. [PMID: 39408862 PMCID: PMC11476611 DOI: 10.3390/ijms251910535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Progress made by the medical community in increasing lifespans comes with the costs of increasing the incidence and prevalence of age-related diseases, neurodegenerative ones included. Aging is associated with a series of morphological changes at the tissue and cellular levels in the brain, as well as impairments in signaling pathways and gene transcription, which lead to synaptic dysfunction and cognitive decline. Although we are not able to pinpoint the exact differences between healthy aging and neurodegeneration, research increasingly highlights the involvement of neuroinflammation and chronic systemic inflammation (inflammaging) in the development of age-associated impairments via a series of pathogenic cascades, triggered by dysfunctions of the circadian clock, gut dysbiosis, immunosenescence, or impaired cholinergic signaling. In addition, gender differences in the susceptibility and course of neurodegeneration that appear to be mediated by glial cells emphasize the need for future research in this area and an individualized therapeutic approach. Although rejuvenation research is still in its very early infancy, accumulated knowledge on the various signaling pathways involved in promoting cellular senescence opens the perspective of interfering with these pathways and preventing or delaying senescence.
Collapse
Affiliation(s)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Alexander Cristian
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Vlad Octavian Hogea
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | | | | |
Collapse
|
2
|
Wen J, Satyanarayanan SK, Li A, Yan L, Zhao Z, Yuan Q, Su KP, Su H. Unraveling the impact of Omega-3 polyunsaturated fatty acids on blood-brain barrier (BBB) integrity and glymphatic function. Brain Behav Immun 2024; 115:335-355. [PMID: 37914102 DOI: 10.1016/j.bbi.2023.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/05/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023] Open
Abstract
Alzheimer's disease (AD) and other forms of dementia represent major public health challenges but effective therapeutic options are limited. Pathological brain aging is associated with microvascular changes and impaired clearance systems. The application of omega-3 polyunsaturated fatty acids (n-3 or omega-3 PUFAs) is one of the most promising nutritional interventions in neurodegenerative disorders from epidemiological data, clinical and pre-clinical studies. As essential components of neuronal membranes, n-3 PUFAs have shown neuroprotection and anti-inflammatory effects, as well as modulatory effects through microvascular pathophysiology, amyloid-beta (Aβ) clearance and glymphatic pathways. This review meticulously explores these underlying mechanisms that contribute to the beneficial effects of n-3 PUFAs against AD and dementia, synthesizing evidence from both animal and interventional studies.
Collapse
Affiliation(s)
- Jing Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Senthil Kumaran Satyanarayanan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong
| | - Ang Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Ziai Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau
| | - Qiuju Yuan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong
| | - Kuan-Pin Su
- An-Nan Hospital, China Medical University, Tainan, Taiwan; Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan; Mind-Body Interface Research Center (MBI-Lab), China Medical University Hospital, Taichung, Taiwan.
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau.
| |
Collapse
|
3
|
Galizzi G, Di Carlo M. Mitochondrial DNA and Inflammation in Alzheimer's Disease. Curr Issues Mol Biol 2023; 45:8586-8606. [PMID: 37998717 PMCID: PMC10670154 DOI: 10.3390/cimb45110540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/15/2023] [Accepted: 10/20/2023] [Indexed: 11/25/2023] Open
Abstract
Mitochondrial dysfunction and neuroinflammation are implicated in the pathogenesis of most neurodegenerative diseases, such as Alzheimer's disease (AD). In fact, although a growing number of studies show crosstalk between these two processes, there remain numerous gaps in our knowledge of the mechanisms involved, which requires further clarification. On the one hand, mitochondrial dysfunction may lead to the release of mitochondrial damage-associated molecular patterns (mtDAMPs) which are recognized by microglial immune receptors and contribute to neuroinflammation progression. On the other hand, inflammatory molecules released by glial cells can influence and regulate mitochondrial function. A deeper understanding of these mechanisms may help identify biomarkers and molecular targets useful for the treatment of neurodegenerative diseases. This review of works published in recent years is focused on the description of the mitochondrial contribution to neuroinflammation and neurodegeneration, with particular attention to mitochondrial DNA (mtDNA) and AD.
Collapse
Affiliation(s)
- Giacoma Galizzi
- Institute for Research and Biomedical Innovation (IRIB), National Research Council (CNR), Via Ugo La Malfa, 153-90146 Palermo, Italy;
| | | |
Collapse
|
4
|
Grayson JM, Short SM, Lee CJ, Park N, Marsac C, Sette A, Lindestam Arlehamn CS, Leng XI, Lockhart SN, Craft S. T cell exhaustion is associated with cognitive status and amyloid accumulation in Alzheimer's disease. Sci Rep 2023; 13:15779. [PMID: 37737298 PMCID: PMC10516910 DOI: 10.1038/s41598-023-42708-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023] Open
Abstract
Studies over the last 100 years have suggested a link between inflammation, infectious disease, and Alzheimer's Disease (AD). Understanding how the immune system changes during the development of AD may facilitate new treatments. Here, we studied an aging cohort who had been assessed for AD pathology with amyloid positron emission tomography and cognitive testing, and conducted high dimensional flow cytometry on peripheral blood mononuclear and cerebrospinal fluid cells. Participants were assigned a classification of being amyloid negative cognitively normal, amyloid positive cognitively normal (APCN), or amyloid positive mild cognitive impairment (APMCI), an early stage of AD. We observed major alterations in the peripheral innate immune system including increased myeloid and plasmacytoid dendritic cells in the blood of APMCI participants. When the adaptive immune system was examined, amyloid positive participants, regardless of cognitive status, had increased CD3+ T cells. Further analyses of CD4+ and CD8+ T cells revealed that APMCI participants had an increase in more differentiated phenotype T cells, such as effector memory and effector memory CD45RA expressing (TEMRA), compared to those with normal cognition. When T cell function was measured, we observed that T cells from APCN participants had increased IFNγ+GzB- producing cells compared to the other participants. In contrast, we demonstrate that APMCI participants had a major increase in T cells that lacked cytokine production following restimulation and expressed increased levels of PD-1 and Tox, suggesting these are exhausted cells. Rejuvenation of these cells may provide a potential treatment for AD.
Collapse
Affiliation(s)
- Jason M Grayson
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 525 Wake Forest Biotech Place, 525 Patterson Avenue Room 2N051, Winston-Salem, NC, 27101, USA.
| | - Samantha M Short
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 525 Wake Forest Biotech Place, 525 Patterson Avenue Room 2N051, Winston-Salem, NC, 27101, USA
| | - C Jiah Lee
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 525 Wake Forest Biotech Place, 525 Patterson Avenue Room 2N051, Winston-Salem, NC, 27101, USA
| | - Nuri Park
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 525 Wake Forest Biotech Place, 525 Patterson Avenue Room 2N051, Winston-Salem, NC, 27101, USA
| | - Caitlyn Marsac
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, 525 Wake Forest Biotech Place, 525 Patterson Avenue Room 2N051, Winston-Salem, NC, 27101, USA
| | - Alessandro Sette
- La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | | | - Xiaoyan I Leng
- Department of Biostatistics and Data Science, One Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Samuel N Lockhart
- Department of Internal Medicine-Geriatrics, One Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Suzanne Craft
- Department of Internal Medicine-Geriatrics, One Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| |
Collapse
|
5
|
Xia Y, Xiao Y, Wang ZH, Liu X, Alam AM, Haran JP, McCormick BA, Shu X, Wang X, Ye K. Bacteroides Fragilis in the gut microbiomes of Alzheimer's disease activates microglia and triggers pathogenesis in neuronal C/EBPβ transgenic mice. Nat Commun 2023; 14:5471. [PMID: 37673907 PMCID: PMC10482867 DOI: 10.1038/s41467-023-41283-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
Gut dysbiosis contributes to Alzheimer's disease (AD) pathogenesis, and Bacteroides strains are selectively elevated in AD gut microbiota. However, it remains unknown which Bacteroides species and how their metabolites trigger AD pathologies. Here we show that Bacteroides fragilis and their metabolites 12-hydroxy-heptadecatrienoic acid (12-HHTrE) and Prostaglandin E2 (PGE2) activate microglia and induce AD pathogenesis in neuronal C/EBPβ transgenic mice. Recolonization of antibiotics cocktail-pretreated Thy1-C/EBPβ transgenic mice with AD patient fecal samples elicits AD pathologies, associated with C/EBPβ/Asparaginyl endopeptidase (AEP) pathway upregulation, microglia activation, and cognitive disorders compared to mice receiving healthy donors' fecal microbiota transplantation (FMT). Microbial 16S rRNA sequencing analysis shows higher abundance of proinflammatory Bacteroides fragilis in AD-FMT mice. Active components characterization from the sera and brains of the transplanted mice revealed that both 12-HHTrE and PGE2 activate primary microglia, fitting with poly-unsaturated fatty acid (PUFA) metabolites enrichment identified by metabolomics. Strikingly, recolonization with live but not dead Bacteroides fragilis elicited AD pathologies in Thy1-C/EBPβ transgenic mice, so did 12-HHTrE or PGE2 treatment alone. Collectively, our findings support a causal role for Bacteroides fragilis and the PUFA metabolites in activating microglia and inducing AD pathologies in Thy1- C/EBPβ transgenic mice.
Collapse
Affiliation(s)
- Yiyuan Xia
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- School of Medicine, Jianghan University, Wuhan, HB, 430056, China
| | - Yifan Xiao
- School of Medicine, Jianghan University, Wuhan, HB, 430056, China
| | - Zhi-Hao Wang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ashfaqul M Alam
- University of Kentucky, Microbiology, Immunology & Molecular Genetics Office - MN 376, Medical Science Building, 800 Rose Street, Lexington, KY, USA
| | - John P Haran
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Beth A McCormick
- Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Xiji Shu
- School of Medicine, Jianghan University, Wuhan, HB, 430056, China.
| | - Xiaochuan Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-innovation Center of Neurodegeneration, Nantong University, Nantong, Jiangsu, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
6
|
Kopp KO, Greer ME, Glotfelty EJ, Hsueh SC, Tweedie D, Kim DS, Reale M, Vargesson N, Greig NH. A New Generation of IMiDs as Treatments for Neuroinflammatory and Neurodegenerative Disorders. Biomolecules 2023; 13:biom13050747. [PMID: 37238617 DOI: 10.3390/biom13050747] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
The immunomodulatory imide drug (IMiD) class, which includes the founding drug member thalidomide and later generation drugs, lenalidomide and pomalidomide, has dramatically improved the clinical treatment of specific cancers, such as multiple myeloma, and it combines potent anticancer and anti-inflammatory actions. These actions, in large part, are mediated by IMiD binding to the human protein cereblon that forms a critical component of the E3 ubiquitin ligase complex. This complex ubiquitinates and thereby regulates the levels of multiple endogenous proteins. However, IMiD-cereblon binding modifies cereblon's normal targeted protein degradation towards a new set of neosubstrates that underlies the favorable pharmacological action of classical IMiDs, but also their adverse actions-in particular, their teratogenicity. The ability of classical IMiDs to reduce the synthesis of key proinflammatory cytokines, especially TNF-α levels, makes them potentially valuable to reposition as drugs to mitigate inflammatory-associated conditions and, particularly, neurological disorders driven by an excessive neuroinflammatory element, as occurs in traumatic brain injury, Alzheimer's and Parkinson's diseases, and ischemic stroke. The teratogenic and anticancer actions of classical IMiDs are substantial liabilities for effective drugs in these disorders and can theoretically be dialed out of the drug class. We review a select series of novel IMiDs designed to avoid binding with human cereblon and/or evade degradation of downstream neosubstrates considered to underpin the adverse actions of thalidomide-like drugs. These novel non-classical IMiDs hold potential as new medications for erythema nodosum leprosum (ENL), a painful inflammatory skin condition associated with Hansen's disease for which thalidomide remains widely used, and, in particular, as a new treatment strategy for neurodegenerative disorders in which neuroinflammation is a key component.
Collapse
Affiliation(s)
- Katherine O Kopp
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| | - Margaret E Greer
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
- Faculty of Medicine, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Elliot J Glotfelty
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Shih-Chang Hsueh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| | - Dong Seok Kim
- Aevisbio Inc., Gaithersburg, MD 20878, USA
- Aevis Bio Inc., Daejeon 34141, Republic of Korea
| | - Marcella Reale
- Department of Innovative Technologies in Medicine and Dentistry, G. d'Annunzio University of Chieti and Pescara, 66100 Chieti, Italy
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| |
Collapse
|
7
|
Rege SV, Teichert A, Masumi J, Dhande OS, Harish R, Higgins BW, Lopez Y, Akrapongpisak L, Hackbart H, Caryotakis S, Leone DP, Szoke B, Hannestad J, Nikolich K, Braithwaite SP, Minami SS. CCR3 plays a role in murine age-related cognitive changes and T-cell infiltration into the brain. Commun Biol 2023; 6:292. [PMID: 36934154 PMCID: PMC10024715 DOI: 10.1038/s42003-023-04665-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 03/06/2023] [Indexed: 03/20/2023] Open
Abstract
Targeting immune-mediated, age-related, biology has the potential to be a transformative therapeutic strategy. However, the redundant nature of the multiple cytokines that change with aging requires identification of a master downstream regulator to successfully exert therapeutic efficacy. Here, we discovered CCR3 as a prime candidate, and inhibition of CCR3 has pro-cognitive benefits in mice, but these benefits are not driven by an obvious direct action on central nervous system (CNS)-resident cells. Instead, CCR3-expressing T cells in the periphery that are modulated in aging inhibit infiltration of these T cells across the blood-brain barrier and reduce neuroinflammation. The axis of CCR3-expressing T cells influencing crosstalk from periphery to brain provides a therapeutically tractable link. These findings indicate the broad therapeutic potential of CCR3 inhibition in a spectrum of neuroinflammatory diseases of aging.
Collapse
|
8
|
HSV-1 cellular model reveals links between aggresome formation and early step of Alzheimer's disease. Transl Psychiatry 2023; 13:86. [PMID: 36898995 PMCID: PMC10006237 DOI: 10.1038/s41398-023-02376-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
Many studies highlight the potential link between the chronic degenerative Alzheimer's disease and the infection by the herpes simplex virus type-1 (HSV-1). However, the molecular mechanisms making possible this HSV-1-dependent process remain to be understood. Using neuronal cells expressing the wild type form of amyloid precursor protein (APP) infected by HSV-1, we characterized a representative cellular model of the early stage of the sporadic form of the disease and unraveled a molecular mechanism sustaining this HSV-1- Alzheimer's disease interplay. Here, we show that HSV-1 induces caspase-dependent production of the 42 amino-acid long amyloid peptide (Aβ42) oligomers followed by their accumulation in neuronal cells. Aβ42 oligomers and activated caspase 3 (casp3A) concentrate into intracytoplasmic structures observed in Alzheimer's disease neuronal cells called aggresomes. This casp3A accumulation in aggresomes during HSV-1 infection limits the execution of apoptosis until its term, similarly to an abortosis-like event occurring in Alzheimer's disease neuronal cells patients. Indeed, this particular HSV-1 driven cellular context, representative of early stages of the disease, sustains a failed apoptosis mechanism that could explain the chronic amplification of Aβ42 production characteristic of Alzheimer's disease patients. Finally, we show that combination of flurbiprofen, a non-steroidal anti-inflammatory drug (NSAID), with caspase inhibitor reduced drastically HSV-1-induced Aβ42 oligomers production. This provided mechanistic insights supporting the conclusion of clinical trials showing that NSAIDs reduced Alzheimer's disease incidence in early stage of the disease. Therefore, from our study we propose that caspase-dependent production of Aβ42 oligomers together with the abortosis-like event represents a vicious circle in early Alzheimer's disease stages leading to a chronic amplification of Aβ42 oligomers that contributes to the establishment of degenerative disorder like Alzheimer's disease in patients infected by HSV-1. Interestingly this process could be targeted by an association of NSAID with caspase inhibitors.
Collapse
|
9
|
Andjelkovic AV, Situ M, Citalan-Madrid AF, Stamatovic SM, Xiang J, Keep RF. Blood-Brain Barrier Dysfunction in Normal Aging and Neurodegeneration: Mechanisms, Impact, and Treatments. Stroke 2023; 54:661-672. [PMID: 36848419 PMCID: PMC9993074 DOI: 10.1161/strokeaha.122.040578] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Cerebral endothelial cells and their linking tight junctions form a unique, dynamic and multi-functional interface, the blood-brain barrier (BBB). The endothelium is regulated by perivascular cells and components forming the neurovascular unit. This review examines BBB and neurovascular unit changes in normal aging and in neurodegenerative disorders, particularly focusing on Alzheimer disease, cerebral amyloid angiopathy and vascular dementia. Increasing evidence indicates BBB dysfunction contributes to neurodegeneration. Mechanisms underlying BBB dysfunction are outlined (endothelium and neurovascular unit mediated) as is the BBB as a therapeutic target including increasing the uptake of systemically delivered therapeutics across the BBB, enhancing clearance of potential neurotoxic compounds via the BBB, and preventing BBB dysfunction. Finally, a need for novel biomarkers of BBB dysfunction is addressed.
Collapse
Affiliation(s)
- Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor MI, USA
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor MI, USA
| | - Muyu Situ
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor MI, USA
| | | | | | - Jianming Xiang
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor MI, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor MI, USA
| |
Collapse
|
10
|
Kumar A, Sudevan ST, Nair AS, Singh AK, Kumar S, Jose J, Behl T, Mangalathillam S, Mathew B, Kim H. Current and Future Nano-Carrier-Based Approaches in the Treatment of Alzheimer's Disease. Brain Sci 2023; 13:brainsci13020213. [PMID: 36831756 PMCID: PMC9953820 DOI: 10.3390/brainsci13020213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
It is a very alarming situation for the globe because 55 million humans are estimated to be affected by Alzheimer's disease (AD) worldwide, and still it is increasing at the rapid speed of 10 million cases per year worldwide. This is an urgent reminder for better research and treatment due to the unavailability of a permanent medication for neurodegenerative disorders like AD. The lack of drugs for neurodegenerative disorder treatment is due to the complexity of the structure of the brain, mainly due to blood-brain barrier, because blood-brain drug molecules must enter the brain compartment. There are several novel and conventional formulation approaches that can be employed for the transportation of drug molecules to the target site in the brain, such as oral, intravenous, gene delivery, surgically implanted intraventricular catheter, nasal and liposomal hydrogels, and repurposing old drugs. A drug's lipophilicity influences metabolic activity in addition to membrane permeability because lipophilic substances have a higher affinity for metabolic enzymes. As a result, the higher a drug's lipophilicity is, the higher its permeability and metabolic clearance. AD is currently incurable, and the medicines available merely cure the symptoms or slow the illness's progression. In the next 20 years, the World Health Organization (WHO) predicts that neurodegenerative illnesses affecting motor function will become the second-leading cause of mortality. The current article provides a brief overview of recent advances in brain drug delivery for AD therapy.
Collapse
Affiliation(s)
- Astik Kumar
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Sachithra Thazhathuveedu Sudevan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Aathira Sujathan Nair
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Ashutosh Kumar Singh
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Sunil Kumar
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Science, NITTE Deemed to be University, Mangalore 575018, India
| | - Tapan Behl
- School of Health Science and Technology, University of Petroleum and Energy Studies, Dehradun 248007, India
| | - Sabitha Mangalathillam
- Department of Pharmaceutics, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682041, India
- Correspondence: (S.M.); or (B.M.); (H.K.)
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, AIMS Health Sciences Campus, Amrita Vishwa Vidyapeetham, Kochi 682041, India
- Correspondence: (S.M.); or (B.M.); (H.K.)
| | - Hoon Kim
- Department of Pharmacy, and Research Institute of Life Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
- Correspondence: (S.M.); or (B.M.); (H.K.)
| |
Collapse
|
11
|
Andronie-Cioara FL, Ardelean AI, Nistor-Cseppento CD, Jurcau A, Jurcau MC, Pascalau N, Marcu F. Molecular Mechanisms of Neuroinflammation in Aging and Alzheimer's Disease Progression. Int J Mol Sci 2023; 24:ijms24031869. [PMID: 36768235 PMCID: PMC9915182 DOI: 10.3390/ijms24031869] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/01/2023] [Accepted: 01/11/2023] [Indexed: 01/20/2023] Open
Abstract
Aging is the most prominent risk factor for late-onset Alzheimer's disease. Aging associates with a chronic inflammatory state both in the periphery and in the central nervous system, the evidence thereof and the mechanisms leading to chronic neuroinflammation being discussed. Nonetheless, neuroinflammation is significantly enhanced by the accumulation of amyloid beta and accelerates the progression of Alzheimer's disease through various pathways discussed in the present review. Decades of clinical trials targeting the 2 abnormal proteins in Alzheimer's disease, amyloid beta and tau, led to many failures. As such, targeting neuroinflammation via different strategies could prove a valuable therapeutic strategy, although much research is still needed to identify the appropriate time window. Active research focusing on identifying early biomarkers could help translating these novel strategies from bench to bedside.
Collapse
Affiliation(s)
- Felicia Liana Andronie-Cioara
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Adriana Ioana Ardelean
- Department of Preclinical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Carmen Delia Nistor-Cseppento
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
- Correspondence: (C.D.N.-C.); (N.P.)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | | | - Nicoleta Pascalau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
- Correspondence: (C.D.N.-C.); (N.P.)
| | - Florin Marcu
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
12
|
Kim J, Kim YK. Molecular Imaging of Neuroinflammation in Alzheimer's Disease and Mild Cognitive Impairment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:301-326. [PMID: 36949316 DOI: 10.1007/978-981-19-7376-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent neurocognitive disorder. Due to the ineffectiveness of treatments targeting the amyloid cascade, molecular biomarkers for neuroinflammation are attracting attention with increasing knowledge about the role of neuroinflammation in the pathogenesis of AD. This chapter will explore the results of studies using molecular imaging for diagnosing AD and mild cognitive impairment (MCI). Because it is critical to interpreting the data to understand which substances are targeted in molecular imaging, this chapter will discuss the two most significant targets, microglia and astrocytes, as well as the best-known radioligands for each. Then, neuroimaging results with PET neuroinflammation imaging will be reviewed for AD and MCI. Although a growing body of evidence has suggested that these molecular imaging biomarkers for neuroinflammation may have a role in the diagnosis of AD and MCI, the findings are inconsistent or cross-sectional, which indicates that it is difficult to apply the contents in practice due to the need for additional study. In particular, because the results of multiple interventions targeting neuroinflammation were inconclusive, molecular imaging markers for neuroinflammation can be used in combination with conventional markers to select appropriate patients for early intervention for neuroinflammation rather than as a single marker.
Collapse
Affiliation(s)
- Junhyung Kim
- Department of Psychiatry, Korea University College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Ansan, Republic of Korea.
| |
Collapse
|
13
|
Walker JM, Dehkordi SK, Schaffert J, Goette W, White CL, Richardson TE, Zare H. The Spectrum of Alzheimer-Type Pathology in Cognitively Normal Individuals. J Alzheimers Dis 2023; 91:683-695. [PMID: 36502330 PMCID: PMC11184733 DOI: 10.3233/jad-220898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The strongest risk factor for the development of Alzheimer's disease (AD) is age. The progression of Braak stage and Thal phase with age has been demonstrated. However, prior studies did not include cognitive status. OBJECTIVE We set out to define normative values for Alzheimer-type pathologic changes in individuals without cognitive decline, and then define levels that would qualify them to be resistant to or resilient against these changes. METHODS Utilizing neuropathology data obtained from the National Alzheimer's Coordinating Center (NACC), we demonstrate the age-related progression of Alzheimer-type pathologic changes in cognitively normal individuals (CDR = 0, n = 542). With plots generated from these data, we establish standard lines that may be utilized to measure the extent to which an individual's Alzheimer-type pathology varies from the estimated normal range of pathology. RESULTS Although Braak stage and Thal phase progressively increase with age in cognitively normal individuals, the Consortium to Establish a Registry for Alzheimer's Disease neuritic plaque score and Alzheimer's disease neuropathologic change remain at low levels. CONCLUSION These findings suggest that an increasing burden of neuritic plaques is a strong predictor of cognitive decline, whereas, neurofibrillary degeneration and amyloid-β (diffuse) plaque deposition, both to some degree, are normal pathologic changes of aging that occur in almost all individuals regardless of cognitive status. Furthermore, we have defined the amount of neuropathologic change in cognitively normal individuals that would qualify them to be "resilient" against the pathology (significantly above the normative values for age, but still cognitively normal) or "resistant" to the development of pathology (significantly below the normative values for age).
Collapse
Affiliation(s)
- Jamie M. Walker
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Shiva Kazempour Dehkordi
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jeff Schaffert
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - William Goette
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Charles L. White
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Timothy E. Richardson
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Habil Zare
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
14
|
Song J, Jia Y, Li J, Ding R, Yuan Y, Cai J, Su Y, Hua Q, Zhang Z. LiuweiDihuang improved cognitive functions in SAMP8 mice by inhibiting COX-2 expression and subsequent neuroinflammation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 296:115491. [PMID: 35752263 DOI: 10.1016/j.jep.2022.115491] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE LiuweiDihuang (LW) pills was mainly used to treatment of children's fontanelle incomplete closure, enuresis and nervous system development delays and other diseases.Following the deepening of pharmacological research, LW has a good effect on neurological diseases include senile dementia. However, the neuroprotection mechanism of LW on Alzheimer's disease (AD) through regulation of inflammation remains unclear. AIM OF THE STUDY Here, we aimed to explore the effects and mechanism of LW on learning and memory deficits in SAMP8 mice. MATERIALS AND METHODS Mice aged 6 months were treated with LW for 2 months and BV2, C6 and HT22 cells were treated with LW pharmaceutic serum and Lipopolysaccharide (LPS) continuously. Then, cognitive tests were performed, including the Morris water maze and Y maze tests. The mRNA level of cyclooxygenase 2 (COX-2) and pro-inflammatory factors (IL-1β, IL-6 and TNF-α) were examined in cells and the cortex and hippocampus by quantitative RT-PCR. The expression of postsynaptic density protein 95, synaptophysin and various inflammatory factors were detected in the cortex and hippocampus by Western blot. Furthermore, Ionized calcium binding adapter molecule 1, glial fibrillary acidic protein and Aβ were examined in the brain of AD mice by immunofluorescence staining and immunohistochemistry. And synaptic loss and neuronal ultrastructure were observed by transmission electron microscope. RESULTS We found that LW suppressed LPS-induced COX-2 expression in vitro. Importantly, LW dramatically improved spatial learning and memory in SAMP8 mice through inhibiting Aβ accumulation and restoring structural synaptic integrity. Furthermore, LW inhibited the glial activation and neuroinflammation (COX-2, IL-1β, IL-6 and TNF-α) in the cortex and hippocampus of SAMP8 mice. CONCLUSION Taken together, the present data not only indicated that LW is an effective agent on improving the learning and memory deficits through mitigating neuroinflammation but highlighted the LW can be a potential therapeutic drug for AD therapy.
Collapse
Affiliation(s)
- Junying Song
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Henan Engineering Research Center for Prevention and Treatment of Neurodegenerative Diseases, Zhengzhou, 450046, PR China; Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Yaquan Jia
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Henan Engineering Research Center for Prevention and Treatment of Neurodegenerative Diseases, Zhengzhou, 450046, PR China
| | - Junlin Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Rui Ding
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Yong Yuan
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Henan Engineering Research Center for Prevention and Treatment of Neurodegenerative Diseases, Zhengzhou, 450046, PR China
| | - Ju Cai
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Medical College, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China
| | - Yunfang Su
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Henan Engineering Research Center for Prevention and Treatment of Neurodegenerative Diseases, Zhengzhou, 450046, PR China
| | - Qian Hua
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China
| | - Zhenqiang Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, PR China; Henan Engineering Research Center for Prevention and Treatment of Neurodegenerative Diseases, Zhengzhou, 450046, PR China.
| |
Collapse
|
15
|
Trinh PNH, Baltos JA, Hellyer SD, May LT, Gregory KJ. Adenosine receptor signalling in Alzheimer’s disease. Purinergic Signal 2022; 18:359-381. [PMID: 35870032 PMCID: PMC9391555 DOI: 10.1007/s11302-022-09883-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/02/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common dementia in the elderly and its increasing prevalence presents treatment challenges. Despite a better understanding of the disease, the current mainstay of treatment cannot modify pathogenesis or effectively address the associated cognitive and memory deficits. Emerging evidence suggests adenosine G protein-coupled receptors (GPCRs) are promising therapeutic targets for Alzheimer’s disease. The adenosine A1 and A2A receptors are expressed in the human brain and have a proposed involvement in the pathogenesis of dementia. Targeting these receptors preclinically can mitigate pathogenic β-amyloid and tau neurotoxicity whilst improving cognition and memory. In this review, we provide an accessible summary of the literature on Alzheimer’s disease and the therapeutic potential of A1 and A2A receptors. Although there are no available medicines targeting these receptors approved for treating dementia, we provide insights into some novel strategies, including allosterism and the targeting of oligomers, which may increase drug discovery success and enhance the therapeutic response.
Collapse
Affiliation(s)
- Phuc N. H. Trinh
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
| | - Shane D. Hellyer
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
| | - Lauren T. May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
| | - Karen J. Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052 Australia
- Department of Pharmacology, Monash University, Parkville, VIC 3052 Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Parkville, 3052 Australia
| |
Collapse
|
16
|
Nehra G, Bauer B, Hartz AMS. Blood-brain barrier leakage in Alzheimer's disease: From discovery to clinical relevance. Pharmacol Ther 2022; 234:108119. [PMID: 35108575 PMCID: PMC9107516 DOI: 10.1016/j.pharmthera.2022.108119] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. AD brain pathology starts decades before the onset of clinical symptoms. One early pathological hallmark is blood-brain barrier dysfunction characterized by barrier leakage and associated with cognitive decline. In this review, we summarize the existing literature on the extent and clinical relevance of barrier leakage in AD. First, we focus on AD animal models and their susceptibility to barrier leakage based on age and genetic background. Second, we re-examine barrier dysfunction in clinical and postmortem studies, summarize changes that lead to barrier leakage in patients and highlight the clinical relevance of barrier leakage in AD. Third, we summarize signaling mechanisms that link barrier leakage to neurodegeneration and cognitive decline in AD. Finally, we discuss clinical relevance and potential therapeutic strategies and provide future perspectives on investigating barrier leakage in AD. Identifying mechanistic steps underlying barrier leakage has the potential to unravel new targets that can be used to develop novel therapeutic strategies to repair barrier leakage and slow cognitive decline in AD and AD-related dementias.
Collapse
Affiliation(s)
- Geetika Nehra
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Bjoern Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
17
|
Long-term low-dose acetylsalicylic use shows protective potential for the development of both vascular dementia and Alzheimer's disease in patients with coronary heart disease but not in other individuals from the general population: results from two large cohort studies. Alzheimers Res Ther 2022; 14:75. [PMID: 35624487 PMCID: PMC9145441 DOI: 10.1186/s13195-022-01017-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/12/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND No population-based cohort study investigated a potential inverse association between long-term low-dose acetylsalicylic acid (ASA) use and all-cause dementia and its two most common sub-types Alzheimer's disease (AD) and vascular dementia (VD) so far. METHODS Cox regression models with inverse probability of treatment weighting to model the underlying cardiovascular risk were used to assess the associations of low-dose ASA use with all-cause dementia, AD, and VD incidence in community-dwelling older adults from the German ESTHER study (N = 5258) and the UK Biobank (N = 305,394). Inclusion criteria were age of 55 years or older and completed drug assessment. Meta-analyses of the individual participant data from the two prospective cohort studies were performed. RESULTS Four hundred seventy-six cases of all-cause dementia, 157 cases of AD, and 183 cases of VD were diagnosed over a median of 14.3 years of follow-up in ESTHER. In the UK Biobank, 5584 participants were diagnosed with all-cause dementia, 2029 with AD, and 1437 with VD over a median of 11.6 years. The meta-analysis of both cohorts revealed a weak reduction in hazards for all-cause dementia (hazard ratio (HR) [95% confidence interval (CI)]: 0.96 [0.93 to 0.99]). The strongest protective effect of low-dose ASA was observed in participants with coronary heart disease (CHD) in both cohorts, and a significant interaction was detected. In particular, in meta-analysis, a 31% reduction in hazard for AD, 69% for VD and 34% for all-cause dementia were observed (HR [95% CI]: 0.69 [0.59 to 0.80], 0.31 [0.27 to 0.35], 0.46 [0.42 to 0.50], respectively). Furthermore, compared to non-users, users of low-dose ASA for 10 years or longer (who likely use it because they have CHD or a related diagnosis putting them at an increased risk for cardiovascular events) demonstrated a strong protective effect on all dementia outcomes, especially for VD (HR [95% CI]: 0.48 [0.42 to 0.56]) whereas no protective associations were observed with shorter low-dose ASA use. CONCLUSIONS The protective potential of low-dose ASA for all-cause dementia, AD, and VD seems to strongly depend on pre-existing CHD and the willingness of patients to take it for a minimum of ten years.
Collapse
|
18
|
Machhi J, Yeapuri P, Markovic M, Patel M, Yan W, Lu Y, Cohen JD, Hasan M, Abdelmoaty MM, Zhou Y, Xiong H, Wang X, Mosley RL, Gendelman HE, Kevadiya BD. Europium-Doped Cerium Oxide Nanoparticles for Microglial Amyloid Beta Clearance and Homeostasis. ACS Chem Neurosci 2022; 13:1232-1244. [PMID: 35312284 PMCID: PMC9227977 DOI: 10.1021/acschemneuro.1c00847] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder. Pathologically, the disease is characterized by the deposition of amyloid beta (Aβ) plaques and the presence of neurofibrillary tangles. These drive microglia neuroinflammation and consequent neurodegeneration. While the means to affect Aβ plaque accumulation pharmacologically was achieved, how it affects disease outcomes remains uncertain. Cerium oxide (CeO2) reduces Aβ plaques, oxidative stress, inflammation, and AD signs and symptoms. In particular, CeO2 nanoparticles (CeO2NPs) induce free-radical-scavenging and cell protective intracellular signaling. This can ameliorate the pathobiology of an AD-affected brain. To investigate whether CeO2NPs affect microglia neurotoxic responses, a novel formulation of europium-doped CeO2NPs (EuCeO2NPs) was synthesized. We then tested EuCeO2NPs for its ability to generate cellular immune homeostasis in AD models. EuCeO2NPs attenuated microglia BV2 inflammatory activities after Aβ1-42 exposure by increasing the cells' phagocytic and Aβ degradation activities. These were associated with increases in the expression of the CD36 scavenger receptor. EuCeO2NPs facilitated Aβ endolysosomal trafficking and abrogated microglial inflammatory responses. We posit that EuCeO2NPs may be developed as an AD immunomodulator.
Collapse
Affiliation(s)
- Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Pravin Yeapuri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Milica Markovic
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Milankumar Patel
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Wenhui Yan
- Department of Pharmacology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi 710061, China
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Jacob D. Cohen
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Mai Mohamed Abdelmoaty
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Giza 12622, Egypt
| | - You Zhou
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Huangui Xiong
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Xinglong Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Bhavesh D. Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
19
|
Mishra A, Wang Y, Yin F, Vitali F, Rodgers KE, Soto M, Mosconi L, Wang T, Brinton RD. A tale of two systems: Lessons learned from female mid-life aging with implications for Alzheimer's prevention & treatment. Ageing Res Rev 2022; 74:101542. [PMID: 34929348 PMCID: PMC8884386 DOI: 10.1016/j.arr.2021.101542] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 12/05/2021] [Accepted: 12/13/2021] [Indexed: 02/03/2023]
Abstract
Neurological aging is frequently viewed as a linear process of decline, whereas in reality, it is a dynamic non-linear process. The dynamic nature of neurological aging is exemplified during midlife in the female brain. To investigate fundamental mechanisms of midlife aging that underlie risk for development of Alzheimer's disease (AD) in late life, we investigated the brain at greatest risk for the disease, the aging female brain. Outcomes of our research indicate that mid-life aging in the female is characterized by the emergence of three phases: early chronological (pre-menopause), endocrinological (peri-menopause) and late chronological (post-menopause) aging. The endocrinological aging program is sandwiched between early and late chronological aging. Throughout the three stages of midlife aging, two systems of biology, metabolic and immune, are tightly integrated through a network of signaling cascades. The network of signaling between these two systems of biology underlie an orchestrated sequence of adaptative starvation responses that shift the brain from near exclusive dependence on a single fuel, glucose, to utilization of an auxiliary fuel derived from lipids, ketone bodies. The dismantling of the estrogen control of glucose metabolism during mid-life aging is a critical contributor to the shift in fuel systems and emergence of dynamic neuroimmune phenotype. The shift in fuel reliance, puts the largest reservoir of local fatty acids, white matter, at risk for catabolism as a source of lipids to generate ketone bodies through astrocytic beta oxidation. APOE4 genotype accelerates the tipping point for emergence of the bioenergetic crisis. While outcomes derived from research conducted in the female brain are not directly translatable to the male brain, the questions addressed in a female centric program of research are directly applicable to investigation of the male brain. Like females, males with AD exhibit deficits in the bioenergetic system of the brain, activation of the immune system and hallmark Alzheimer's pathologies. The drivers and trajectory of mechanisms underlying neurodegeneration in the male brain will undoubtedly share common aspects with the female in addition to factors unique to the male. Preclinical and clinical evidence indicate that midlife endocrine aging can also be a transitional bridge to autoimmune disorders. Collectively, the data indicate that endocrinological aging is a critical period "tipping point" in midlife which can initiate emergence of the prodromal stage of late-onset-Alzheimer's disease. Interventions that target both immune and metabolic shifts that occur during midlife aging have the potential to alter the trajectory of Alzheimer's risk in late life. Further, to achieve precision medicine for AD, chromosomal sex is a critical variable to consider along with APOE genotype, other genetic risk factors and stage of disease.
Collapse
Affiliation(s)
- Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Fei Yin
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Francesca Vitali
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Kathleen E Rodgers
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Maira Soto
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Tian Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Roberta D Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA.
| |
Collapse
|
20
|
4R-cembranoid protects neuronal cells from oxygen-glucose deprivation by modulating microglial cell activation. Brain Res Bull 2022; 179:74-82. [PMID: 34942325 PMCID: PMC8849140 DOI: 10.1016/j.brainresbull.2021.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 02/03/2023]
Abstract
As major immune responsive cells in the central nervous system (CNS), activated microglia can present pro-inflammatory M1 phenotype aggravating the neuronal injury or anti-inflammatory M2 phenotype providing neuroprotection and promoting neuronal survival in neurodegenerative diseases. In this study, we demonstrated that a compound, 4R-cembranoid (4R, 1S, 2E, 4R, 6R,-7E, 11E-2, 7, 11-cembratriene-4, 6-diol cembranoids) promoted M2 phenotype while attenuated M1 phenotype in N9 cells, a microglial cell line. Following Lipopolysaccharides (LPS) or Oxygen-glucose deprivation (OGD) treatment, the N9 cells treated by 1 µM 4R showed an increased Arginase-1 (Arg1, a M2 marker) expression and a reduced inducible nitric oxide synthase (iNOS, M1 marker) expression. In addition, the conditioned medium of 4R-treated post-OGD N9 cells protected neuro2a cells, a neuronal cell line, from OGD-induced injury. The viability of neuro2a cells in OGD condition was increased by 54.5% after treated with the conditioned medium of 4R-treated post-OGD N9 cells. Furthermore, we demonstrated the protective mechanism of 4R was associated with a decreased TNF-α release and an increased IL-10 release from N9 cells. In conclusion, our study demonstrated that the neuroprotective effects of 4R were through the regulation of microglial activation by promoting the protective M2 activation and inhibiting the damaging M1 activation. Therefore, the findings of this study suggest that 4R could be a promising lead structure for the development of drugs for the treatment of ischemic stroke and other neurodegenerative diseases with an inflammatory component involved.
Collapse
|
21
|
Onaolapo OJ, Olofinnade AT, Ojo FO, Onaolapo AY. Neuroinflammation and Oxidative Stress in Alzheimer's Disease; Can Nutraceuticals and Functional Foods Come to the Rescue? Antiinflamm Antiallergy Agents Med Chem 2022; 21:75-89. [PMID: 36043770 DOI: 10.2174/1871523021666220815151559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/17/2022] [Accepted: 06/29/2022] [Indexed: 06/15/2023]
Abstract
Alzheimer's disease (AD), the most prevalent form of age-related dementia, is typified by progressive memory loss and spatial awareness with personality changes. The increasing socioeconomic burden associated with AD has made it a focus of extensive research. Ample scientific evidence supports the role of neuroinflammation and oxidative stress in AD pathophysiology, and there is increasing research into the possible role of anti-inflammatory and antioxidative agents as disease modifying therapies. While, the result of numerous preclinical studies has demonstrated the benefits of anti-inflammatory agents, these benefits however have not been replicated in clinical trials, necessitating a further search for more promising anti-inflammatory agents. Current understanding highlights the role of diet in the development of neuroinflammation and oxidative stress, as well as the importance of dietary interventions and lifestyle modifications in mitigating them. The current narrative review examines scientific literature for evidence of the roles (if any) of dietary components, nutraceuticals and functional foods in the prevention or management of AD. It also examines how diet/ dietary components could modulate oxidative stress/inflammatory mediators and pathways that are crucial to the pathogenesis and/or progression of AD.
Collapse
Affiliation(s)
- Olakunle J Onaolapo
- Department of Pharmacology, Behavioural Neuroscience Unit, Neuropharmacology Subdivision, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Anthony T Olofinnade
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Clinical Sciences, College of Medicine, Lagos State University, Ikeja, Lagos State, Nigeria
| | - Folusho O Ojo
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Adejoke Y Onaolapo
- Department of Anatomy, Behavioural Neuroscience Unit, Neurobiology Subdivision, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
22
|
McFarland KN, Chakrabarty P. Microglia in Alzheimer's Disease: a Key Player in the Transition Between Homeostasis and Pathogenesis. Neurotherapeutics 2022; 19:186-208. [PMID: 35286658 PMCID: PMC9130399 DOI: 10.1007/s13311-021-01179-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2021] [Indexed: 02/07/2023] Open
Abstract
Immune activation accompanies the development of proteinopathy in the brains of Alzheimer's dementia patients. Evolving from the long-held viewpoint that immune activation triggers the pathological trajectory in Alzheimer's disease, there is accumulating evidence now that microglial activation is neither pro-amyloidogenic nor just a simple reactive process to the proteinopathy. Preclinical studies highlight an interesting aspect of immunity, i.e., spurring immune system activity may be beneficial under certain circumstances. Indeed, a dynamic evolving relationship between different activation states of the immune system and its neuronal neighbors is thought to regulate overall brain organ health in both healthy aging and progression of Alzheimer's dementia. A new premise evolving from genome, transcriptome, and proteome data is that there might be at least two major phases of immune activation that accompany the pathological trajectory in Alzheimer's disease. Though activation on a chronic scale will certainly lead to neurodegeneration, this emerging knowledge of a potential beneficial aspect of immune activation allows us to form holistic insights into when, where, and how much immune system activity would need to be tuned to impact the Alzheimer's neurodegenerative cascade. Even with the trove of recently emerging -omics data from patients and preclinical models, how microglial phenotypes are functionally related to the transition of a healthy aging brain towards progressive degenerative state remains unknown. A deeper understanding of the synergism between microglial functional states and brain organ health could help us discover newer interventions and therapies that enable us to address the current paucity of disease-modifying therapies in Alzheimer's disease.
Collapse
Affiliation(s)
- Karen N McFarland
- Department of Neurology, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA.
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
23
|
Mavroudis I, Chowdhury R, Petridis F, Karantali E, Chatzikonstantinou S, Balmus IM, Luca IS, Ciobica A, Kazis D. YKL-40 as a Potential Biomarker for the Differential Diagnosis of Alzheimer's Disease. MEDICINA (KAUNAS, LITHUANIA) 2021; 58:medicina58010060. [PMID: 35056368 PMCID: PMC8777884 DOI: 10.3390/medicina58010060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder, associated with extensive neuronal loss, dendritic and synaptic changes resulting in significant cognitive impairment. An increased number of studies have given rise to the neuroinflammatory hypothesis in AD. It is widely accepted that AD brains show chronic inflammation, probably triggered by the presence of insoluble amyloid beta deposits and neurofibrillary tangles (NFT) and is also related to the activation of neuronal death cascade. In the present study we aimed to investigate the role of YKL-40 levels in the cerebrospinal fluid (CSF) in the diagnosis of AD, and to discuss whether there are further potential roles of this protein in the management and treatment of AD. We conducted an online search on PubMed, Web of Science, and the Cochrane library databases from 1990 to 2021. The quantitative analysis showed that the levels of YKL-40 were significantly higher in Alzheimer’s disease compared to controls, to mild cognitive impairment (MCI) AD (MCI-AD) and to stable MCI. They were also increased in MCI-AD compared to stable MCI. The present study shows that the CSF levels of YKL-40 could be potentially used as a biomarker for the prognosis of mild cognitive impairment and the likelihood of progression to AD, as well as for the differential diagnosis between AD and MCI.
Collapse
Affiliation(s)
- Ioannis Mavroudis
- Department of Neurology, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK; (I.M.); (R.C.)
| | - Rumana Chowdhury
- Department of Neurology, Leeds Teaching Hospitals, NHS Trust, Leeds LS2 9JT, UK; (I.M.); (R.C.)
| | - Foivos Petridis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| | - Eleni Karantali
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| | - Symela Chatzikonstantinou
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| | - Ioana Miruna Balmus
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, ”Alexandru Ioan Cuza” University of Iasi, Alexandru Lapsuneanu Street, No. 26, 700057 Iasi, Romania;
| | - Iuliana Simona Luca
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, No. 20A, 700505 Iasi, Romania
- Correspondence: (I.S.L.); (A.C.)
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, No. 20A, 700505 Iasi, Romania
- Correspondence: (I.S.L.); (A.C.)
| | - Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (F.P.); (E.K.); (S.C.); (D.K.)
| |
Collapse
|
24
|
Peters R, Breitner J, James S, Jicha GA, Meyer P, Richards M, Smith AD, Yassine HN, Abner E, Hainsworth AH, Kehoe PG, Beckett N, Weber C, Anderson C, Anstey KJ, Dodge HH. Dementia risk reduction: why haven't the pharmacological risk reduction trials worked? An in-depth exploration of seven established risk factors. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12202. [PMID: 34934803 PMCID: PMC8655351 DOI: 10.1002/trc2.12202] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/03/2021] [Accepted: 06/18/2021] [Indexed: 12/21/2022]
Abstract
Identifying the leading health and lifestyle factors for the risk of incident dementia and Alzheimer's disease has yet to translate to risk reduction. To understand why, we examined the discrepancies between observational and clinical trial evidence for seven modifiable risk factors: type 2 diabetes, dyslipidemia, hypertension, estrogens, inflammation, omega-3 fatty acids, and hyperhomocysteinemia. Sample heterogeneity and paucity of intervention details (dose, timing, formulation) were common themes. Epidemiological evidence is more mature for some interventions (eg, non-steroidal anti-inflammatory drugs [NSAIDs]) than others. Trial data are promising for anti-hypertensives and B vitamin supplementation. Taken together, these risk factors highlight a future need for more targeted sample selection in clinical trials, a better understanding of interventions, and deeper analysis of existing data.
Collapse
Affiliation(s)
- Ruth Peters
- Neuroscience ResearchSydneyNew South WalesAustralia
- Department of Psychology University of New South WalesSydneyNew South WalesAustralia
| | - John Breitner
- Douglas Hospital Research Center and McGill UniversityQuebecCanada
| | - Sarah James
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | | | - Pierre‐Francois Meyer
- Center for Studies on the Prevention of Alzheimer's Disease (PREVENT‐AD)VerdunQuebecCanada
| | - Marcus Richards
- MRC Unit for Lifelong Health and Ageing at UCLUniversity College LondonLondonUK
| | - A. David Smith
- OPTIMADepartment of PharmacologyUniversity of OxfordOxfordUK
| | - Hussein N. Yassine
- Departments of Medicine and NeurologyUniversity of Southern CaliforniaCaliforniaUSA
| | - Erin Abner
- University of KentuckyLexingtonKentuckyUSA
| | - Atticus H. Hainsworth
- Molecular and Clinical Sciences Research InstituteSt GeorgesUniversity of LondonLondonUK
- Department of NeurologySt George's HospitalLondonUK
| | | | | | | | - Craig Anderson
- The George Institute for Global HealthSydneyNew South WalesAustralia
| | - Kaarin J. Anstey
- Neuroscience ResearchSydneyNew South WalesAustralia
- Department of Psychology University of New South WalesSydneyNew South WalesAustralia
| | | |
Collapse
|
25
|
Banik A, Amaradhi R, Lee D, Sau M, Wang W, Dingledine R, Ganesh T. Prostaglandin EP2 receptor antagonist ameliorates neuroinflammation in a two-hit mouse model of Alzheimer's disease. J Neuroinflammation 2021; 18:273. [PMID: 34801055 PMCID: PMC8605573 DOI: 10.1186/s12974-021-02297-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/14/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) causes substantial medical and societal burden with no therapies ameliorating cognitive deficits. Centralized pathologies involving amyloids, neurofibrillary tangles, and neuroinflammatory pathways are being investigated to identify disease-modifying targets for AD. Cyclooxygenase-2 (COX-2) is one of the potential neuroinflammatory agents involved in AD progression. However, chronic use of COX-2 inhibitors in patients produced adverse cardiovascular effects. We asked whether inhibition of EP2 receptors, downstream of the COX-2 signaling pathway, can ameliorate neuroinflammation in AD brains in presence or absence of a secondary inflammatory stimuli. METHODS We treated 5xFAD mice and their non-transgenic (nTg) littermates in presence or absence of lipopolysaccharide (LPS) with an EP2 antagonist (TG11-77.HCl). In cohort 1, nTg (no-hit) or 5xFAD (single-hit-genetic) mice were treated with vehicle or TG11-77.HCl for 12 weeks. In cohort 2, nTg (single-hit-environmental) and 5xFAD mice (two-hit) were administered LPS (0.5 mg/kg/week) and treated with vehicle or TG11-77.HCl for 8 weeks. RESULTS Complete blood count analysis showed that LPS induced anemia of inflammation in both groups in cohort 2. There was no adverse effect of LPS or EP2 antagonist on body weight throughout the treatment. In the neocortex isolated from the two-hit cohort of females, but not males, the elevated mRNA levels of proinflammatory mediators (IL-1β, TNF, IL-6, CCL2, EP2), glial markers (IBA1, GFAP, CD11b, S110B), and glial proteins were significantly reduced by EP2 antagonist treatment. Intriguingly, the EP2 antagonist had no effect on either of the single-hit cohorts. There was a modest increase in amyloid-plaque deposition upon EP2 antagonist treatment in the two-hit female brains, but not in the single-hit genetic female cohort. CONCLUSION These results reveal a potential neuroinflammatory role for EP2 in the two-hit 5xFAD mouse model. A selective EP2 antagonist reduces inflammation only in female AD mice subjected to a second inflammatory insult.
Collapse
Affiliation(s)
- Avijit Banik
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Radhika Amaradhi
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Daniel Lee
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Michael Sau
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Wenyi Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
26
|
Czapski GA, Strosznajder JB. Glutamate and GABA in Microglia-Neuron Cross-Talk in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms222111677. [PMID: 34769106 PMCID: PMC8584169 DOI: 10.3390/ijms222111677] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/18/2022] Open
Abstract
The physiological balance between excitation and inhibition in the brain is significantly affected in Alzheimer’s disease (AD). Several neuroactive compounds and their signaling pathways through various types of receptors are crucial in brain homeostasis, among them glutamate and γ-aminobutyric acid (GABA). Activation of microglial receptors regulates the immunological response of these cells, which in AD could be neuroprotective or neurotoxic. The novel research approaches revealed the complexity of microglial function, including the interplay with other cells during neuroinflammation and in the AD brain. The purpose of this review is to describe the role of several proteins and multiple receptors on microglia and neurons, and their involvement in a communication network between cells that could lead to different metabolic loops and cell death/survival. Our review is focused on the role of glutamatergic, GABAergic signaling in microglia–neuronal cross-talk in AD and neuroinflammation. Moreover, the significance of AD-related neurotoxic proteins in glutamate/GABA-mediated dialogue between microglia and neurons was analyzed in search of novel targets in neuroprotection, and advanced pharmacological approaches.
Collapse
|
27
|
Garbuz DG, Zatsepina OG, Evgen’ev MB. Beta Amyloid, Tau Protein, and Neuroinflammation: An Attempt to Integrate Different Hypotheses of Alzheimer’s Disease Pathogenesis. Mol Biol 2021. [DOI: 10.1134/s002689332104004x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease that inevitably results in dementia and death. Currently, there are no pathogenetically grounded methods for the prevention and treatment of AD, and all current treatment regimens are symptomatic and unable to significantly delay the development of dementia. The accumulation of β-amyloid peptide (Aβ), which is a spontaneous, aggregation-prone, and neurotoxic product of the processing of signaling protein APP (Amyloid Precursor Protein), in brain tissues, primarily in the hippocampus and the frontal cortex, was for a long time considered the main cause of neurodegenerative changes in AD. However, attempts to treat AD based on decreasing Aβ production and aggregation did not bring significant clinical results. More and more arguments are arising in favor of the fact that the overproduction of Aβ in most cases of AD is not the initial cause, but a concomitant event of pathological processes in the course of the development of sporadic AD. The concept of neuroinflammation has come to the fore, suggesting that inflammatory responses play the leading role in the initiation and development of AD, both in brain tissue and in the periphery. The hypothesis about the key role of neuroinflammation in the pathogenesis of AD opens up new opportunities in the search for ways to treat and prevent this socially significant disease.
Collapse
|
28
|
de Oliveira J, Kucharska E, Garcez ML, Rodrigues MS, Quevedo J, Moreno-Gonzalez I, Budni J. Inflammatory Cascade in Alzheimer's Disease Pathogenesis: A Review of Experimental Findings. Cells 2021; 10:cells10102581. [PMID: 34685563 PMCID: PMC8533897 DOI: 10.3390/cells10102581] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia worldwide. Most AD patients develop the disease in late life, named late onset AD (LOAD). Currently, the most recognized explanation for AD pathology is the amyloid cascade hypothesis. It is assumed that amyloid beta (Aβ) aggregation and deposition are critical pathogenic processes in AD, leading to the formation of amyloid plaques, as well as neurofibrillary tangles, neuronal cell death, synaptic degeneration, and dementia. In LOAD, the causes of Aβ accumulation and neuronal loss are not completely clear. Importantly, the blood–brain barrier (BBB) disruption seems to present an essential role in the induction of neuroinflammation and consequent AD development. In addition, we propose that the systemic inflammation triggered by conditions like metabolic diseases or infections are causative factors of BBB disruption, coexistent inflammatory cascade and, ultimately, the neurodegeneration observed in AD. In this regard, the use of anti-inflammatory molecules could be an interesting strategy to treat, delay or even halt AD onset and progression. Herein, we review the inflammatory cascade and underlying mechanisms involved in AD pathogenesis and revise the anti-inflammatory effects of compounds as emerging therapeutic drugs against AD.
Collapse
Affiliation(s)
- Jade de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90050-000, Brazil; (J.d.O.); (M.S.R.)
| | - Ewa Kucharska
- Faculty of Education, Institute of Educational Sciences, Jesuit University Ignatianum in Krakow, 31-501 Krakow, Poland;
| | - Michelle Lima Garcez
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis 88040-900, Santa Catarina, Brazil;
| | - Matheus Scarpatto Rodrigues
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 90050-000, Brazil; (J.d.O.); (M.S.R.)
| | - João Quevedo
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA;
- Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
- Neuroscience Graduate Program, Graduate School of Biomedical Sciences, MD Anderson Cancer Center, UTHealth, The University of Texas Houston, Houston, TX 77030, USA
- Graduate Program in Health Sciences, Translational Psychiatry Laboratory, University of Southern Santa Catarina (UNESC), Criciuma 88806-000, Brazil
| | - Ines Moreno-Gonzalez
- Department of Cell Biology, Faculty of Sciences, University of Malaga, IBIMA, 29010 Malaga, Spain;
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), 29010 Malaga, Spain
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Josiane Budni
- Programa de Pós-Graduação em Ciências da Saúde, Laboratório de Neurologia Experimental, Universidade do Extremo Sul Catarinense, Criciuma 88806-000, Brazil
- Correspondence: ; Tel.: +55-48431-2539
| |
Collapse
|
29
|
Hayley S, Hakim AM, Albert PR. Depression, dementia and immune dysregulation. Brain 2021; 144:746-760. [PMID: 33279966 DOI: 10.1093/brain/awaa405] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/26/2020] [Accepted: 09/20/2020] [Indexed: 12/17/2022] Open
Abstract
Major depression is a prevalent illness that increases the risk of several neurological conditions. These include stroke, cardiovascular disease, and dementia including Alzheimer's disease. In this review we ask whether certain types of depression and associated loneliness may be a harbinger of cognitive decline and possibly even dementia. We propose that chronic stress and inflammation combine to compromise vascular and brain function. The resulting increases in proinflammatory cytokines and microglial activation drive brain pathology leading to depression and mild cognitive impairment, which may progress to dementia. We present evidence that by treating the inflammatory changes, depression can be reversed in many cases. Importantly, there is evidence that anti-inflammatory and antidepressant treatments may reduce or prevent dementia in people with depression. Thus, we propose a model in which chronic stress and inflammation combine to increase brain permeability and cytokine production. This leads to microglial activation, white matter damage, neuronal and glial cell loss. This is first manifest as depression and mild cognitive impairment, but can eventually evolve into dementia. Further research may identify clinical subgroups with inflammatory depression at risk for dementia. It would then be possible to address in clinical trials whether effective treatment of the depression can delay the onset of dementia.
Collapse
Affiliation(s)
- Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Antoine M Hakim
- Ottawa Hospital Research Institute (Neuroscience), uOttawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), uOttawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
30
|
McDade E, Llibre-Guerra JJ, Holtzman DM, Morris JC, Bateman RJ. The informed road map to prevention of Alzheimer Disease: A call to arms. Mol Neurodegener 2021; 16:49. [PMID: 34289882 PMCID: PMC8293489 DOI: 10.1186/s13024-021-00467-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/10/2021] [Indexed: 12/31/2022] Open
Abstract
Alzheimer disease (AD) prevention trials hold the promise to delay or prevent cognitive decline and dementia onset by intervening before significant neuronal damage occurs. In recent years, the first AD prevention trials have launched and are yielding important findings on the biology of targeting asymptomatic AD pathology. However, there are limitations that impact the design of these prevention trials, including the translation of animal models that recapitulate key stages and multiple pathological aspects of the human disease, missing target validation in asymptomatic disease, uncertain causality of the association of pathophysiologic changes with cognitive and clinical symptoms, and limited biomarker validation for novel targets. The field is accelerating advancements in key areas including the development of highly specific and quantitative biomarker measures for AD pathology, increasing our understanding of the course and relationship of amyloid and tau pathology in asymptomatic through symptomatic stages, and the development of powerful interventions that can slow or reverse AD amyloid pathology. We review the current status of prevention trials and propose key areas of needed research as a call to basic and translational scientists to accelerate AD prevention. Specifically, we review (1) sporadic and dominantly inherited primary and secondary AD prevention trials, (2) proposed targets, mechanisms, and drugs including the amyloid, tau, and inflammatory pathways and combination treatments, (3) the need for more appropriate prevention animal models and experiments, and (4) biomarkers and outcome measures needed to design human asymptomatic prevention trials. We conclude with actions needed to effectively move prevention targets and trials forward.
Collapse
Affiliation(s)
- Eric McDade
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Avenue, Campus Box, St Louis, MO 8111 USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Dominantly Inherited Alzheimer’s Network Trials Unit, St. Louis, MO 63110 USA
| | - Jorge J. Llibre-Guerra
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Avenue, Campus Box, St Louis, MO 8111 USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Dominantly Inherited Alzheimer’s Network Trials Unit, St. Louis, MO 63110 USA
| | - David M. Holtzman
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Avenue, Campus Box, St Louis, MO 8111 USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Dominantly Inherited Alzheimer’s Network Trials Unit, St. Louis, MO 63110 USA
| | - John C. Morris
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Avenue, Campus Box, St Louis, MO 8111 USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Dominantly Inherited Alzheimer’s Network Trials Unit, St. Louis, MO 63110 USA
| | - Randall J. Bateman
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Avenue, Campus Box, St Louis, MO 8111 USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Dominantly Inherited Alzheimer’s Network Trials Unit, St. Louis, MO 63110 USA
| |
Collapse
|
31
|
Yuen SC, Liang X, Zhu H, Jia Y, Leung SW. Prediction of differentially expressed microRNAs in blood as potential biomarkers for Alzheimer's disease by meta-analysis and adaptive boosting ensemble learning. Alzheimers Res Ther 2021; 13:126. [PMID: 34243793 PMCID: PMC8272278 DOI: 10.1186/s13195-021-00862-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 06/17/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Blood circulating microRNAs that are specific for Alzheimer's disease (AD) can be identified from differentially expressed microRNAs (DEmiRNAs). However, non-reproducible and inconsistent reports of DEmiRNAs hinder biomarker development. The most reliable DEmiRNAs can be identified by meta-analysis. To enrich the pool of DEmiRNAs for potential AD biomarkers, we used a machine learning method called adaptive boosting for miRNA disease association (ABMDA) to identify eligible candidates that share similar characteristics with the DEmiRNAs identified from meta-analysis. This study aimed to identify blood circulating DEmiRNAs as potential AD biomarkers by augmenting meta-analysis with the ABMDA ensemble learning method. METHODS Studies on DEmiRNAs and their dysregulation states were corroborated with one another by meta-analysis based on a random-effects model. DEmiRNAs identified by meta-analysis were collected as positive examples of miRNA-AD pairs for ABMDA ensemble learning. ABMDA identified similar DEmiRNAs according to a set of predefined criteria. The biological significance of all resulting DEmiRNAs was determined by their target genes according to pathway enrichment analyses. The target genes common to both meta-analysis- and ABMDA-identified DEmiRNAs were collected to construct a network to investigate their biological functions. RESULTS A systematic database search found 7841 studies for an extensive meta-analysis, covering 54 independent comparisons of 47 differential miRNA expression studies, and identified 18 reliable DEmiRNAs. ABMDA ensemble learning was conducted based on the meta-analysis results and the Human MicroRNA Disease Database, which identified 10 additional AD-related DEmiRNAs. These 28 DEmiRNAs and their dysregulated pathways were related to neuroinflammation. The dysregulated pathway related to neuronal cell cycle re-entry (CCR) was the only statistically significant pathway of the ABMDA-identified DEmiRNAs. In the biological network constructed from 1865 common target genes of the identified DEmiRNAs, the multiple core ubiquitin-proteasome system, that is involved in neuroinflammation and CCR, was highly connected. CONCLUSION This study identified 28 DEmiRNAs as potential AD biomarkers in blood, by meta-analysis and ABMDA ensemble learning in tandem. The DEmiRNAs identified by meta-analysis and ABMDA were significantly related to neuroinflammation, and the ABMDA-identified DEmiRNAs were related to neuronal CCR.
Collapse
Affiliation(s)
- Sze Chung Yuen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078 Macao China
| | - Xiaonan Liang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078 Macao China
| | - Hongmei Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078 Macao China
| | - Yongliang Jia
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, 999078 Macao China
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Siu-wai Leung
- Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen, China
- Edinburgh Bayes Centre for AI Research in Shenzhen, College of Science and Engineering, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
32
|
Kaduševičius E. Novel Applications of NSAIDs: Insight and Future Perspectives in Cardiovascular, Neurodegenerative, Diabetes and Cancer Disease Therapy. Int J Mol Sci 2021; 22:6637. [PMID: 34205719 PMCID: PMC8235426 DOI: 10.3390/ijms22126637] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 01/22/2023] Open
Abstract
Once it became clear that inflammation takes place in the modulation of different degenerative disease including neurodegenerative, cardiovascular, diabetes and cancer the researchers has started intensive programs evaluating potential role of non-steroidal anti-inflammatory drugs (NSAIDs) in the prevention or therapy of these diseases. This review discusses the novel mechanism of action of NSAIDs and its potential use in the pharmacotherapy of neurodegenerative, cardiovascular, diabetes and cancer diseases. Many different molecular and cellular factors which are not yet fully understood play an important role in the pathogenesis of inflammation, axonal damage, demyelination, atherosclerosis, carcinogenesis thus further NSAID studies for a new potential indications based on precise pharmacotherapy model are warranted since NSAIDs are a heterogeneous group of medicines with relative different pharmacokinetics and pharmacodynamics profiles. Hopefully the new data from studies will fill in the gap between experimental and clinical results and translate our knowledge into successful disease therapy.
Collapse
Affiliation(s)
- Edmundas Kaduševičius
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, 9 A. Mickeviciaus Street, LT-44307 Kaunas, Lithuania
| |
Collapse
|
33
|
Morgan DG, Mielke MM. Knowledge gaps in Alzheimer's disease immune biomarker research. Alzheimers Dement 2021; 17:2030-2042. [PMID: 33984178 DOI: 10.1002/alz.12342] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 11/09/2022]
Abstract
Considerable evidence has accumulated implicating a role for immune mechanisms in moderating the pathology in Alzheimer's disease dementia. However, the appropriate therapeutic target, the appropriate direction of manipulation, and the stage of disease at which to begin treatment remain unanswered questions. Part of the challenge derives from the absence of any selective pressure to develop a coordinated beneficial immune response to severe neural injury in adults. Thus, immune responses to the prevailing stimuli are likely to contain both beneficial and detrimental components. Knowledge gaps include: (1) how a biomarker change relates to the underlying biology, (2) the degree to which pathological stage group differences reflect a response to pathology versus trait differences among individuals regulating risk of developing pathology, (3) the degree to which biomarker levels are predictive of subsequent changes in pathology and/or cognition, and (4) experimental manipulations in model systems to determine whether differences in immune biomarkers are causally related to pathology.
Collapse
Affiliation(s)
- David G Morgan
- Alzheimer's Alliance, Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, Michigan, USA
| | - Michelle M Mielke
- Division of Epidemiology, Department of Health Sciences Research, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
34
|
Abstract
One of the best strategies for healthy brain aging is regular aerobic exercise. Commonly studied "anti-aging" compounds may mimic some effects of exercise on the brain, but novel approaches that target energy-sensing pathways similar to exercise probably will be more effective in this context. We review evidence in support of this hypothesis by focusing on biological hallmarks of brain aging.
Collapse
|
35
|
Long QH, Wu YG, He LL, Ding L, Tan AH, Shi HY, Wang P. Suan-Zao-Ren Decoction ameliorates synaptic plasticity through inhibition of the Aβ deposition and JAK2/STAT3 signaling pathway in AD model of APP/PS1 transgenic mice. Chin Med 2021; 16:14. [PMID: 33478552 PMCID: PMC7818567 DOI: 10.1186/s13020-021-00425-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/08/2021] [Indexed: 01/23/2023] Open
Abstract
Background Suan-Zao-Ren Decoction (SZRD) has been widely used to treat neurological illnesses, including dementia, insomnia and depression. However, the mechanisms underlying SZRD’s improvement in cognitive function remain unclear. In this study, we examined SZRD’s effect on APP/PS1 transgenic mice and mechanisms associated with SZRD’s action in alleviating neuroinflammation and improving synaptic plasticity. Methods
The APP/PS1 mice were treated with different dosages of SZRD (12.96 and 25.92 g/kg/day, in L-SZRD and H-SZRD groups, respectively) for 4 weeks. Morris water maze was conducted to determine changes in behaviors of the mice after the treatment. Meanwhile, in the samples of the hippocampus, Nissl staining and Golgi-Cox staining were used to detect synaptic plasticity. ELISA was applied to assess the expression levels of Aβ1−40 and Aβ1−42 in the hippocampus of mice. Western blot (WB) was employed to test the protein expression level of Aβ1−42, APP, ADAM10, BACE1, PS1, IDE, IBA1, GFAP, PSD95 and SYN, as well as the expressions of JAK2, STAT3 and their phosphorylation patterns to detect the involvement of JAK2/STAT3 pathway. Besides, we examined the serum and hippocampal contents of IL-1β, IL-6 and TNF-α through ELISA. Results Compared to the APP/PS1 mice without any treatment, SZRD, especially the L-SZRD, significantly ameliorated cognitive impairment of the APP/PS1 mice with decreases in the loss of neurons and Aβ plaque deposition as well as improvement of synaptic plasticity in the hippocampus (P < 0.05 or 0.01). Also, SZRD, in particular, the L-SZRD markedly inhibited the serum and hippocampal concentrations of IL-6, IL-1β and TNF-α, while reducing the expression of p-JAK2-Tyr1007 and p-STAT3-Tyr705 in the hippocampus of the APP/PS1 mice (P < 0.05 or 0.01). Conclusions The SZRD, especially the L-SZRD, may improve the cognitive impairment and ameliorate the neural degeneration in APP/PS1 transgenic mice through inhibiting Aβ accumulation and neuroinflammation via the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Qing-Hua Long
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei, China
| | - Yong-Gui Wu
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei, China
| | - Li-Ling He
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei, China
| | - Li Ding
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei, China
| | - Ai-Hua Tan
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei, China
| | - He-Yuan Shi
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei, China.
| | - Ping Wang
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei, China.
| |
Collapse
|
36
|
Severini C, Barbato C, Di Certo MG, Gabanella F, Petrella C, Di Stadio A, de Vincentiis M, Polimeni A, Ralli M, Greco A. Alzheimer's Disease: New Concepts on the Role of Autoimmunity and NLRP3 Inflammasome in the Pathogenesis of the Disease. Curr Neuropharmacol 2021; 19:498-512. [PMID: 32564756 PMCID: PMC8206463 DOI: 10.2174/1570159x18666200621204546] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 05/27/2020] [Accepted: 06/17/2020] [Indexed: 01/14/2023] Open
Abstract
Alzheimer's disease (AD), recognized as the most common neurodegenerative disorder, is clinically characterized by the presence of extracellular beta-amyloid (Aβ) plaques and by intracellular neurofibrillary tau tangles, accompanied by glial activation and neuroinflammation. Increasing evidence suggests that self-misfolded proteins stimulate an immune response mediated by glial cells, inducing the release of inflammatory mediators and the recruitment of peripheral macrophages into the brain, which in turn aggravate AD pathology. The present review aims to update the current knowledge on the role of autoimmunity and neuroinflammation in the pathogenesis of the disease, indicating a new target for therapeutic intervention. We mainly focused on the NLRP3 microglial inflammasome as a critical factor in stimulating innate immune responses, thus sustaining chronic inflammation. Additionally, we discussed the involvement of the NLRP3 inflammasome in the gut-brain axis. Direct targeting of the NLRP3 inflammasome and the associated receptors could be a potential pharmacological strategy since its inhibition would selectively reduce AD neuroinflammation.
Collapse
Affiliation(s)
- Cinzia Severini
- Address correspondence to this author at the Institute of Biochemistry and Cell Biology, National Research Council of Italy, Viale del Policlinico, 155, 00161 Rome, Italy; E-mail:
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Streit WJ, Khoshbouei H, Bechmann I. The Role of Microglia in Sporadic Alzheimer's Disease. J Alzheimers Dis 2020; 79:961-968. [PMID: 33361603 DOI: 10.3233/jad-201248] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Microglia constitute the brain's immune system and their involvement in Alzheimer's disease has been discussed. Commonly, and in line with the amyloid/neuroinflammation cascade hypothesis, microglia have been portrayed as potentially dangerous immune effector cells thought to be overactivated by amyloid and producing neurotoxic inflammatory mediators that lead to neurofibrillary degeneration. We disagree with this theory and offer as an alternative the microglial dysfunction theory stating that microglia become impaired in their normally neuroprotective roles because of aging, i.e., they become senescent and aging neurons degenerate because they lack the needed microglial support for their survival. Thus, while the amyloid cascade theory relies primarily on genetic data, the dysfunction theory incorporates aging as a critical etiological factor. Aging is the greatest risk factor for the sporadic (late-onset) and most common form of Alzheimer's disease, where fully penetrant genetic mutations are absent. In this review, we lay out and discuss the human evidence that supports senescent microglial dysfunction and conflicts with the amyloid/neuroinflammation idea.
Collapse
Affiliation(s)
- Wolfgang J Streit
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | | | - Ingo Bechmann
- Anatomical Institute, University of Leipzig, Leipzig, Germany
| |
Collapse
|
38
|
Manji Z, Rojas A, Wang W, Dingledine R, Varvel NH, Ganesh T. 5xFAD Mice Display Sex-Dependent Inflammatory Gene Induction During the Prodromal Stage of Alzheimer's Disease. J Alzheimers Dis 2020; 70:1259-1274. [PMID: 31322556 DOI: 10.3233/jad-180678] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) pathology consists of extracellular deposits of amyloid-β peptides (Aβ) and intracellular neurofibrillary tangles. These pathological alterations are accompanied by a neuroinflammatory response consisting of increased expression of inflammatory mediators. An anti-inflammatory strategy designed to prevent or delay the development of AD would benefit from knowing when neuroinflammation appears in the transgenic models during prodromal disease stages relative to Aβ pathology. We investigated the expression patterns of inflammatory mediators in the brain of 5xFAD mice in comparison to development of Aβ deposition. Expression changes in inflammatory mediators and glial markers are more robust in female mice starting at three months of age, in contrast to males in which there is no clear trend through five months. Female and male 5xFAD mice also displayed an age-dependent increase in cortical Aβ deposition congruent with neuroinflammation. Thus, in the 5xFAD mouse model of AD, administration of an anti-inflammatory agent would be most efficacious when administered before three months of age.
Collapse
Affiliation(s)
- Zahra Manji
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Asheebo Rojas
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Wenyi Wang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas H Varvel
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
39
|
Thomas AJ, Hamilton CA, Donaghy PC, Martin-Ruiz C, Morris CM, Barnett N, Olsen K, Taylor JP, O'Brien JT. Prospective longitudinal evaluation of cytokines in mild cognitive impairment due to AD and Lewy body disease. Int J Geriatr Psychiatry 2020; 35:1250-1259. [PMID: 32557792 DOI: 10.1002/gps.5365] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/04/2020] [Accepted: 06/13/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES We conducted a prospective longitudinal study of plasma cytokines during the Mild Cognitive Impairment (MCI) stage of Lewy body disease and Alzheimer's disease, hypothesizing that cytokine levels would decrease over time and that this would be correlated with decline in cognition. METHODS Older (≥60) people with MCI were recruited from memory services in healthcare trusts in North East England, UK. MCI was diagnosed as due to Alzheimer's disease (MCI-AD) or Lewy body disease (MCI-LB). Baseline and repeat annual clinical and cognitive assessments were undertaken and plasma samples were obtained at the same time. Cytokine assays were performed on all samples using the Meso Scale Discovery V-Plex Plus Proinflammatory Panel 1, which included IFNγ, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13 and TNFα. RESULTS Fifty-six patients (21 MCI-AD, 35 MCI-LB) completed prospective evaluations and provided samples up to 3 years after baseline. Six cytokines (IFNγ, IL-1β, IL-2, IL-4, IL-6 and IL-10) showed highly significant (P < .002) decreases over time. AD and LB did not differ in rate of decrease nor were there any effects related to age or general morbidity. Decrease in five of these cytokines (IFNγ, IL-1β, IL-2, IL-4, and IL-10) was highly correlated with decrease in cognition (P < .003). CONCLUSIONS Peripheral inflammation decreased in both disease groups during MCI suggesting this may be a therapeutic window for future anti-inflammatory agents.
Collapse
Affiliation(s)
- Alan J Thomas
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Calum A Hamilton
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Paul C Donaghy
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Carmen Martin-Ruiz
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Chris M Morris
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Nicola Barnett
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Kirsty Olsen
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
40
|
Mhillaj E, Papi M, Paciello F, Silvestrini A, Rolesi R, Palmieri V, Perini G, Fetoni AR, Trabace L, Mancuso C. Celecoxib Exerts Neuroprotective Effects in β-Amyloid-Treated SH-SY5Y Cells Through the Regulation of Heme Oxygenase-1: Novel Insights for an Old Drug. Front Cell Dev Biol 2020; 8:561179. [PMID: 33134292 PMCID: PMC7550645 DOI: 10.3389/fcell.2020.561179] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/31/2020] [Indexed: 12/25/2022] Open
Abstract
The formation and aggregation of amyloid-β-peptide (Aβ) into soluble and insoluble species represent the pathological hallmarks of Alzheimer’s disease (AD). Over the last few years, however, soluble Aβ (sAβ) prevailed over fibrillar Aβ (fAβ) as determinant of neurotoxicity. One of the main therapeutic strategies for challenging neurodegeneration is to fight against neuroinflammation and prevent free radical-induced damage: in this light, the heme oxygenase/biliverdin reductase (HO/BVR) system is considered a promising drug target. The aim of this work was to investigate whether or not celecoxib (CXB), a selective inhibitor of the pro-inflammatory cyclooxygenase-2, modulates the HO/BVR system and prevents lipid peroxidation in SH-SY5Y neuroblastoma cells. Both sAβ (6.25–50 nM) and fAβ (1.25–50 nM) dose-dependently over-expressed inducible HO (HO-1) after 24 h of incubation, reaching statistical significance at 25 and 6.25 nM, respectively. Interestingly, CXB (1–10 μM, for 1 h) further enhanced Aβ-induced HO-1 expression through the nuclear translocation of the transcriptional factor Nrf2. Furthermore, 10 μM CXB counteracted the Aβ-induced ROS production with a mechanism fully dependent on HO-1 up-regulation; nevertheless, 10 μM CXB significantly counteracted only 25 nM sAβ-induced lipid peroxidation damage in SH-SY5Y neurons by modulating HO-1. Both carbon monoxide (CORM-2, 50 nM) and bilirubin (50 nM) significantly prevented ROS production in Aβ-treated neurons and favored both the slowdown of the growth rate of Aβ oligomers and the decrease in oligomer/fibril final size. In conclusion, these results suggest a novel mechanism through which CXB is neuroprotective in subjects with early AD or mild cognitive impairment.
Collapse
Affiliation(s)
- Emanuela Mhillaj
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Massimiliano Papi
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Fabiola Paciello
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Andrea Silvestrini
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Rolando Rolesi
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Palmieri
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giordano Perini
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Rita Fetoni
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Cesare Mancuso
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
41
|
Pinheiro L, Faustino C. Therapeutic Strategies Targeting Amyloid-β in Alzheimer's Disease. Curr Alzheimer Res 2020; 16:418-452. [PMID: 30907320 DOI: 10.2174/1567205016666190321163438] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/16/2019] [Accepted: 03/17/2019] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder linked to protein misfolding and aggregation. AD is pathologically characterized by senile plaques formed by extracellular Amyloid-β (Aβ) peptide and Intracellular Neurofibrillary Tangles (NFT) formed by hyperphosphorylated tau protein. Extensive synaptic loss and neuronal degeneration are responsible for memory impairment, cognitive decline and behavioral dysfunctions typical of AD. Amyloidosis has been implicated in the depression of acetylcholine synthesis and release, overactivation of N-methyl-D-aspartate (NMDA) receptors and increased intracellular calcium levels that result in excitotoxic neuronal degeneration. Current drugs used in AD treatment are either cholinesterase inhibitors or NMDA receptor antagonists; however, they provide only symptomatic relief and do not alter the progression of the disease. Aβ is the product of Amyloid Precursor Protein (APP) processing after successive cleavage by β- and γ-secretases while APP proteolysis by α-secretase results in non-amyloidogenic products. According to the amyloid cascade hypothesis, Aβ dyshomeostasis results in the accumulation and aggregation of Aβ into soluble oligomers and insoluble fibrils. The former are synaptotoxic and can induce tau hyperphosphorylation while the latter deposit in senile plaques and elicit proinflammatory responses, contributing to oxidative stress, neuronal degeneration and neuroinflammation. Aβ-protein-targeted therapeutic strategies are thus a promising disease-modifying approach for the treatment and prevention of AD. This review summarizes recent findings on Aβ-protein targeted AD drugs, including β-secretase inhibitors, γ-secretase inhibitors and modulators, α-secretase activators, direct inhibitors of Aβ aggregation and immunotherapy targeting Aβ, focusing mainly on those currently under clinical trials.
Collapse
Affiliation(s)
- Lídia Pinheiro
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto 1649-003 Lisboa, Portugal
| | - Célia Faustino
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto 1649-003 Lisboa, Portugal
| |
Collapse
|
42
|
Salech F, Ponce DP, Paula-Lima AC, SanMartin CD, Behrens MI. Nicotinamide, a Poly [ADP-Ribose] Polymerase 1 (PARP-1) Inhibitor, as an Adjunctive Therapy for the Treatment of Alzheimer's Disease. Front Aging Neurosci 2020; 12:255. [PMID: 32903806 PMCID: PMC7438969 DOI: 10.3389/fnagi.2020.00255] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/24/2020] [Indexed: 12/25/2022] Open
Abstract
Nicotinamide (vitamin B3) is a key component in the cellular production of Nicotinamide Adenine Dinucleotide (NAD) and has long been associated with neuronal development, survival and death. Numerous data suggest that nicotinamide may offer therapeutic benefits in neurodegenerative disorders, including Alzheimer’s Disease (AD). Beyond its effect in NAD+ stores, nicotinamide is an inhibitor of Poly [ADP-ribose] polymerase 1 (PARP-1), an enzyme with multiple cellular functions, including regulation of cell death, energy/metabolism and inflammatory response. PARP-1 functions as a DNA repair enzyme but under intense DNA damage depletes the cell of NAD+ and ATP and leads to a non-apoptotic type of cell death called Parthanatos, which has been associated with the pathogenesis of neurodegenerative diseases. Moreover, NAD+ availability might potentially improve mitochondrial function, which is severely impaired in AD. PARP-1 inhibition may also exert a protective effect against neurodegeneration by its action to diminish neuroinflammation and microglial activation which are also implicated in the pathogenesis of AD. Here we discuss the evidence supporting the use of nicotinamide as adjunctive therapy for the treatment of early stages of AD based on the inhibitory effect of nicotinamide on PARP-1 activity. The data support evaluating nicotinamide as an adjunctive treatment for AD at early stages of the disease not only to increase NAD+ stores but as a PARP-1 inhibitor, raising the hypothesis that other PARP-1 inhibitors – drugs that are already approved for breast cancer treatment – might be explored for the treatment of AD.
Collapse
Affiliation(s)
- Felipe Salech
- Centro de Investigación Clínica Avanzada, Facultad de Medicina and Hospital Clínico Universidad de Chile, Santiago, Chile.,Sección de Geriatría Hospital Clínico Universidad de Chile, Santiago, Chile.,Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Daniela P Ponce
- Centro de Investigación Clínica Avanzada, Facultad de Medicina and Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Andrea C Paula-Lima
- Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Biomedical Neuroscience Institute, Facultad of Medicina, Universidad de Chile, Santiago, Chile.,Institute for Research in Dental Sciences, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Carol D SanMartin
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.,Departamento de Neurologiìa y Neurocirugiìa, Hospital Cliìnico Universidad de Chile, Santiago, Chile
| | - María I Behrens
- Centro de Investigación Clínica Avanzada, Facultad de Medicina and Hospital Clínico Universidad de Chile, Santiago, Chile.,Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Departamento de Neurologiìa y Neurocirugiìa, Hospital Cliìnico Universidad de Chile, Santiago, Chile.,Departamento de Neurología y Psiquiatría, Clínica Alemana de Santiago, Santiago, Chile
| |
Collapse
|
43
|
Fernandes A, Tábuas-Pereira M, Duro D, Lima M, Gens H, Santiago B, Durães J, Almeida MR, Leitão MJ, Baldeiras I, Santana I. C-reactive protein as a predictor of mild cognitive impairment conversion into Alzheimer's disease dementia. Exp Gerontol 2020; 138:111004. [PMID: 32561398 DOI: 10.1016/j.exger.2020.111004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 06/10/2020] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND AIMS Increasing evidence suggests that inflammation plays an important role in brain aging and neurodegeneration. Pathological studies demonstrate the presence of C-reactive protein (CRP) in the senile plaques and neurofibrillary tangles in Alzheimer's disease (AD) brain tissue suggesting that CRP may play a role in its neuropathological processes. Some findings suggest that midlife elevations of serum CRP are a risk factor for AD. However, others found lower CRP levels in mild or moderate AD than in controls, suggesting that CRP levels could be different in different stages of disease. We aimed to assess the role of CRP as a predictor of Mild cognitive impairment (MCI) conversion into AD dementia. METHODS We retrospectively reviewed the cohort of MCI patients followed at the Dementia Clinic, Neurology Department of University Hospital of Coimbra. We collected demographical, neuropsychological, genetic and laboratorial variables (including serum CRP measurements at the time of baseline laboratory tests). A Cox regression model was performed adjusted for the collected variables preconsidered to be predictors of dementia and the variable being studied (CRP) to assess for independent predictors of conversion. RESULTS We included 130 patients, 58.5% female, with a mean age of onset of 65.5 ± 9.1 years and age at first assessment of 69.3 ± 8.5 years. The mean CRP was 0.33 ± 0.58 mg/dl. At follow-up (mean, 36.9 ± 27.0 months) 42.3% of MCI patients converted to dementia. Lower CSF Aβ42 (HR = 0.999, 95%CI = [0.997, 1.000], p = 0.015), lower MMSE score (HR = 0.864, 95%CI = [0.510, 1.595], p = 0.008) and lower CRP quartile (HR = 0.597, 95%CI = [0.435, 0.819], p = 0.001) were independent predictors of conversion. CONCLUSION CRP may add information of risk of conversion in MCI patients. Patients with lower CRP levels appear to have a more rapid conversion to AD dementia.
Collapse
Affiliation(s)
- Andreia Fernandes
- Neurology Department, Centro Hospitalar e Universitário de Lisboa Central, Alameda de Santo António dos Capuchos, 1169-050 Lisboa, Portugal.
| | - Miguel Tábuas-Pereira
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal
| | - Diana Duro
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal
| | - Marisa Lima
- Faculty of Psychology and Educational Sciences, University of Coimbra, R. Colégio Novo, 3000-115 Coimbra, Portugal
| | - Helena Gens
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal
| | - Beatriz Santiago
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal
| | - João Durães
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal
| | - Maria Rosário Almeida
- Faculty of Medicine, University of Coimbra, R. Larga, 3004-504 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Maria João Leitão
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Inês Baldeiras
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, R. Larga, 3004-504 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Isabel Santana
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, R. Larga, 3004-504 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| |
Collapse
|
44
|
Potter H, Boyd TD, Clarke P, Pelak VS, Tyler KL. Recruiting the innate immune system with GM-CSF to fight viral diseases, including West Nile Virus encephalitis and COVID-19. F1000Res 2020; 9:345. [PMID: 32704352 PMCID: PMC7359749 DOI: 10.12688/f1000research.23729.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/30/2020] [Indexed: 01/08/2023] Open
Abstract
As the coronavirus disease 2019 (COVID-19) pandemic grows throughout the world, it is imperative that all approaches to ameliorating its effects be investigated, including repurposing drugs that show promise in other diseases. We have been investigating an approach to multiple disorders that involves recruiting the innate immune system to aid the body's healing and regenerative mechanism(s). In the case of West Nile Virus encephalitis and potentially COVID-19, the proposed intervention to stimulate the innate immune system may give the adaptive immune response the necessary time to develop, finish clearing the virus, and provide future immunity. Furthermore, we have found that GM-CSF-induced recruitment of the innate immune system is also able to reverse brain pathology, neuroinflammation and cognitive deficits in mouse models of Alzheimer's disease and Down syndrome, as well as improving cognition in normal aging and in human patients with cognitive deficits due to chemotherapy, both of which exhibit neuroinflammation. Others have shown that GM-CSF is an effective treatment for both bacterial and viral pneumonias, and their associated inflammation, in animals and that it has successfully treated pneumonia-associated Acute Respiratory Distress Syndrome in humans. These and other data strongly suggest that GM-CSF may be an effective treatment for many viral infections, including COVID-19.
Collapse
Affiliation(s)
- Huntington Potter
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA
| | - Timothy D. Boyd
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, Aurora, CO, 80045, USA
| | - Penny Clarke
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Victoria S. Pelak
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado Alzheimer's and Cognition Center, Aurora, CO, 80045, USA
| | - Kenneth L. Tyler
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
45
|
Monobutyrin and Monovalerin Affect Brain Short-Chain Fatty Acid Profiles and Tight-Junction Protein Expression in ApoE-Knockout Rats Fed High-Fat Diets. Nutrients 2020; 12:nu12041202. [PMID: 32344633 PMCID: PMC7230324 DOI: 10.3390/nu12041202] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/13/2020] [Accepted: 04/20/2020] [Indexed: 12/19/2022] Open
Abstract
Monobutyrin (MB) and monovalerin (MV), esters of short-chain fatty acids (SCFAs), have previously been shown to reduce liver cholesterol and inflammation in conventional rats fed high-fat diets. This study explored the potential effects of MB and MV in hypercholesterolemic apolipoprotein E-knockout (ApoE-/-) rats. ApoE-/- rats were fed three high-fat (HF) diets, pure or supplemented with MB or MV (1%), for 5 weeks. One group of conventional rats (C) was also fed the pure high-fat diet and another group of ApoE-/- rats a low-fat (LF) diet. Blood and liver lipids, urinary lactulose/mannitol, SCFAs (blood and brain), tight junction proteins (small intestine and brain), and inflammation-related markers (blood, brain, and liver) were analyzed. MV supplementation elevated serum high-density lipoprotein (HDL) cholesterol and valeric acid concentration (p < 0.05), while the amounts of isovaleric acid in the brain were reduced (p < 0.05). MB increased butyric acid amounts in the brain, while the plasma concentration of interleukin 10 (IL-10) was lowered (p < 0.05). Both MV and MB upregulated the expression of occludin and zonula occludens-1 (ZO-1) in the brain (p < 0.05). Supplementation of MB or MV affected HDL cholesterol, the expression of tight junction proteins, and SCFA profiles. MB and MV may therefore be promising supplements to attenuate lipid metabolic disorders caused by high-fat intake and genetic deficiency.
Collapse
|
46
|
Early intraneuronal amyloid triggers neuron-derived inflammatory signaling in APP transgenic rats and human brain. Proc Natl Acad Sci U S A 2020; 117:6844-6854. [PMID: 32144141 PMCID: PMC7104377 DOI: 10.1073/pnas.1914593117] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
This work provides evidence that soluble and oligomeric amyloid protein stokes neuronal inflammation during the earliest stages of Alzheimer’s disease. Identifying neuron-derived factors that engage the brain’s immune system will provide insight into how vulnerable neurons might interact with other immune cells to propagate cytotoxic signaling cascades and cellular dysfunction during disease development. Chronic inflammation during Alzheimer’s disease (AD) is most often attributed to sustained microglial activation in response to amyloid-β (Aβ) plaque deposits and cell death. However, cytokine release and microgliosis are consistently observed in AD transgenic animal models devoid of such pathologies, bringing into question the underlying processes that may be at play during the earliest AD-related immune response. We propose that this plaque-independent inflammatory reaction originates from neurons burdened with increasing levels of soluble and oligomeric Aβ, which are known to be the most toxic amyloid species within the brain. Laser microdissected neurons extracted from preplaque amyloid precursor protein (APP) transgenic rats were found to produce a variety of potent immune factors, both at the transcript and protein levels. Neuron-derived cytokines correlated with the extent of microglial activation and mobilization, even in the absence of extracellular plaques and cell death. Importantly, we identified an inflammatory profile unique to Aβ-burdened neurons, since neighboring glial cells did not express similar molecules. Moreover, we demonstrate within disease-vulnerable regions of the human brain that a neuron-specific inflammatory response may precede insoluble Aβ plaque and tau tangle formation. Thus, we reveal the Aβ-burdened neuron as a primary proinflammatory agent, implicating the intraneuronal accumulation of Aβ as a significant immunological component in the AD pathogenesis.
Collapse
|
47
|
Neurovascular unit dysregulation, white matter disease, and executive dysfunction: the shared triad of vascular cognitive impairment and Alzheimer disease. GeroScience 2020; 42:445-465. [PMID: 32002785 DOI: 10.1007/s11357-020-00164-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/22/2020] [Indexed: 01/07/2023] Open
Abstract
Executive dysfunction is the most important predictor for loss of independence in dementia. As executive function involves the coordination of distributed cerebral functions, executive function requires healthy white matter. However, white matter is highly vulnerable to cerebrovascular insults, with executive dysfunction being a core feature of vascular cognitive impairment (VCI). At the same time, cerebrovascular pathology, white matter disease, and executive dysfunction are all increasingly recognized as features of Alzheimer disease (AD). Recent studies have characterized the crucial role of glial cells in the pathological changes observed in both VCI and AD. In comorbid VCI and AD, the glial cells of the neurovascular unit (NVU) emerge as important therapeutic targets for the preservation of white matter integrity and executive function. Our synthesis from current research identifies dysregulation of the NVU, white matter disease, and executive dysfunction as a fundamental triad that is common to both VCI and AD. Further study of this triad will be critical for advancing the prevention and management of dementia.
Collapse
|
48
|
Rogers NK, Romero C, SanMartín CD, Ponce DP, Salech F, López MN, Gleisner A, Tempio F, Behrens MI. Inverse Relationship Between Alzheimer’s Disease and Cancer: How Immune Checkpoints Might Explain the Mechanisms Underlying Age-Related Diseases. J Alzheimers Dis 2020; 73:443-454. [DOI: 10.3233/jad-190839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Nicole K. Rogers
- Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Unidad de Paciente Crítico, Instituto de Neurocirugía Asenjo, Santiago, Chile
| | - Cesar Romero
- Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Carol D. SanMartín
- Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Santiago, Chile
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Daniela P. Ponce
- Centro de Investigación Clínica Avanzada (CICA), Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Felipe Salech
- Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Centro de Investigación Clínica Avanzada (CICA), Hospital Clínico Universidad de Chile, Santiago, Chile
- Sección de Geriatría, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Mercedes N. López
- Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alejandra Gleisner
- Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Fabián Tempio
- Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - María I. Behrens
- Departamento de Neurociencia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Santiago, Chile
- Centro de Investigación Clínica Avanzada (CICA), Hospital Clínico Universidad de Chile, Santiago, Chile
- Clínica Alemana de Santiago, Santiago, Chile
| |
Collapse
|
49
|
Tenner AJ. Complement-Mediated Events in Alzheimer's Disease: Mechanisms and Potential Therapeutic Targets. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:306-315. [PMID: 31907273 PMCID: PMC6951444 DOI: 10.4049/jimmunol.1901068] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/18/2019] [Indexed: 12/11/2022]
Abstract
An estimated 5.7 million Americans suffer from Alzheimer's disease in the United States, with no disease-modifying treatments to prevent or treat cognitive deficits associated with the disease. Genome-wide association studies suggest that an enhancement of clearance mechanisms and/or promotion of an anti-inflammatory response may slow or prevent disease progression. Increasing awareness of distinct roles of complement components in normal brain development and function and in neurodegenerative disorders align with complement-mediated responses, and thus, thorough understanding of these molecular pathways is needed to facilitate successful therapeutic design. Both beneficial and detrimental effects of C1q as well as contributions to local inflammation by C5a-C5aR1 signaling in brain highlight the need for precision of therapeutic design. The potential benefit of β-amyloid clearance from the circulation via CR1-mediated mechanisms is also reviewed. Therapies that suppress inflammation while preserving protective effects of complement could be tested now to slow the progression of this debilitating disease.
Collapse
Affiliation(s)
- Andrea J Tenner
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697;
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697;
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697; and
- Institute for Memory Impairment and Neurological Disorders, University of California Irvine, Irvine, CA 92697
| |
Collapse
|
50
|
Streit WJ, Khoshbouei H, Bechmann I. Dystrophic microglia in late-onset Alzheimer's disease. Glia 2020; 68:845-854. [PMID: 31922322 DOI: 10.1002/glia.23782] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 12/23/2019] [Accepted: 12/30/2019] [Indexed: 01/08/2023]
Abstract
Here, we summarize current understanding of functional involvement of microglial cells in the most common neurodegenerative disease to affect humans, which is sporadic or late-onset Alzheimer's disease (LOAD). Our review narrowly focuses on insights obtained from post-mortem neuropathological examinations of human brains paying particular attention to microglia as these cells have long been implicated as pivotal players in the cellular processes that lead to AD-type neurodegeneration. Although complete understanding of the roles played by microglia in AD neurodegeneration remains elusive, our studies thus far have illuminated microglial involvement in LOAD, showing that microglial dystrophy, the morphological manifestation of senescence, can be integrated with other hallmark pathological features of AD, such as intraneuronal neurofibrillary degeneration (NFD) and extracellular deposits of amyloid-beta (Aβ) protein. We have demonstrated an in situ correlation between microglial dystrophy and presence of NFD suggesting that neurodegeneration is secondary to aging-related microglial deterioration, a concept founded on the notion that proper neuronal function is dependent on presence of healthy microglia. Diseased or weakened glia are detrimental for neuronal well-being because their ability to provide neuronal support may be impaired. Our most recent work also links microglial dystrophy with Aβ deposits by showing that there is a chronic, yet futile microglial reaction to insoluble amyloid deposits. This inability of microglia to remove aggregated amyloid (a foreign body) causes microglial exhaustion and thereby exacerbates already ongoing aging-dependent microglial deterioration. An eventual total loss of functional microglia in advanced LOAD promotes widespread NFD, dementia, and brain failure.
Collapse
Affiliation(s)
- Wolfgang J Streit
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida
| | - Ingo Bechmann
- Anatomical Institute, University of Leipzig, Leipzig, Germany
| |
Collapse
|